Fact-checked by Grok 2 weeks ago

NOD-like receptor

NOD-like receptors (NLRs) are a family of cytosolic receptors essential to the , functioning as sensors that detect intracellular pathogen-associated molecular patterns (PAMPs) from microbes and damage-associated molecular patterns (DAMPs) from host cell stress or damage. These receptors, numbering 23 in humans and 34 in mice, enable rapid immune responses by recognizing diverse ligands such as bacterial peptidoglycans (e.g., γ-D-glutamyl-meso-diaminopimelic acid for NOD1 and muramyl for ), toxins, viral components, and endogenous signals like ATP or crystals. Upon ligand binding, NLRs undergo conformational changes that promote self-oligomerization, forming multiprotein complexes known as or nodosomes to initiate downstream signaling. Structurally, NLRs share a conserved tripartite architecture: an N-terminal effector domain (variably a pyrin domain [PYD], caspase activation and recruitment domain [CARD], acidic domain, baculovirus inhibitor of apoptosis repeat [BIR], or unknown in NLRX1), a central nucleotide-binding oligomerization domain (NOD or NACHT) that facilitates ATP-dependent oligomerization, and a C-terminal leucine-rich repeat (LRR) domain responsible for ligand sensing and autoregulation. Exceptions include NLRP1, which has a C-terminal CARD instead of an N-terminal one, and NLRP10, lacking LRRs. Based on the N-terminal domain, NLRs are classified into subfamilies: NLRA (e.g., CIITA, involved in MHC class II expression), NLRB (e.g., NAIP, neuronal apoptosis inhibitor protein), NLRC (e.g., NOD1/NLRC1 and NOD2/NLRC2 for peptidoglycan sensing, NLRC4 for bacterial flagellin and type III secretion systems), NLRP (e.g., NLRP3, the most studied for broad DAMP/PAMP detection), and NLRX (e.g., NLRX1 for mitochondrial antiviral signaling). Evolutionarily, NLRs trace back to plant resistance (R) genes, with expansions through gene duplication in vertebrates, reflecting adaptations to diverse threats. In terms of function, activated NLRs trigger multiple pathways: NLRC subfamily members like NOD1 and activate (NF-κB) and (MAPK) signaling to induce proinflammatory cytokines (e.g., TNF-α, IL-6) and , while also promoting against intracellular . NLRP and NLRC4 inflammasomes recruit adaptor protein ASC and procaspase-1, leading to caspase-1 autoactivation, which cleaves pro-IL-1β and pro-IL-18 into mature forms and gasdermin D for cell death, a lytic process that amplifies . Some NLRs, such as , integrate multiple signals (e.g., potassium efflux, , lysosomal damage) via regulators like NEK7, and can form broader complexes like PANoptosomes involving ZBP1 or AIM2 to drive PANoptosis (, , necroptosis). Beyond immunity, NLRs influence (e.g., NLRP6 in production) and adaptive responses (e.g., NLRC5 in expression). Dysregulation of NLRs contributes to a spectrum of diseases, underscoring their therapeutic potential. Gain-of-function mutations in cause cryopyrin-associated periodic syndromes (CAPS), including familial cold autoinflammatory syndrome and Muckle-Wells syndrome, characterized by excessive IL-1β-driven inflammation. Loss-of-function variants in are strongly associated with , impairing bacterial clearance and leading to chronic intestinal inflammation. Other links include NLRP1 in skin disorders like , NLRC4 in autoinflammatory syndromes with macrophage activation, and NLRP3 hyperactivity in , , , and neurodegenerative diseases like Alzheimer's via chronic sterile inflammation. In infections, NLR deficiencies heighten susceptibility to pathogens like or , while overactivation can exacerbate or severity. Ongoing research targets NLR pathways with inhibitors (e.g., MCC950 for NLRP3) to balance immunity and inflammation.

Overview

Definition and Discovery

NOD-like receptors (NLRs), also known as nucleotide-binding oligomerization domain-like receptors, constitute a family of intracellular receptors (PRRs) that detect pathogen-associated molecular patterns (PAMPs) from invading microbes and damage-associated molecular patterns (DAMPs) from host cells under stress or damage, thereby initiating innate immune responses such as and pathways. These receptors are primarily expressed in immune cells like macrophages and dendritic cells but also in epithelial and other non-immune cells, enabling surveillance of the cytosolic compartment for threats that evade extracellular detection. The discovery of NLRs emerged from early 2000s research on innate immunity, building on the identification of related proteins in the apoptotic machinery. NOD1, the first member, was cloned and characterized in 1999 by Inohara et al. as a protein with a , nucleotide-binding domain (NBD), and , capable of activating and in response to intracellular signals. This was followed by functional studies in 2000 demonstrating NOD1's role in activation through an induced proximity model involving the kinase RICK. NOD2 was identified shortly after in 2001 through positional cloning efforts, with Ogura et al. describing it as a susceptibility gene for via frameshift mutations that impair its function, and Hugot et al. confirming variants associated with the inflammatory bowel disorder. These findings established NLRs as key sensors in host defense and linked their dysregulation to human disease. NLRs exhibit evolutionary conservation across eukaryotes, with foundational homologs in represented by nucleotide-binding site-leucine-rich repeat (NBS-LRR) proteins that mediate resistance to pathogens through similar domain architectures for sensing and signaling. In vertebrates, the NLR family expanded to 23 members in humans and 34 in mice, reflecting adaptations in complex immune systems. This conservation underscores NLRs' ancient role in cytosolic immunity, predating the divergence of plant and animal lineages.

Biological Significance

NOD-like receptors (NLRs) serve as critical intracellular sensors in the , detecting microbial patterns and cellular stress signals to initiate protective responses against pathogens. They play essential roles in host defense by recognizing components from bacteria, viruses, and other invaders, thereby triggering cytokine production and antimicrobial mechanisms that bolster immune . For instance, NLRs such as NOD1 and detect bacterial derivatives, enabling rapid responses to intracellular infections. Additionally, certain NLRs respond to sterile injury signals, such as damage-associated molecular patterns (DAMPs), helping to resolve tissue damage and prevent excessive inflammation. Beyond innate immunity, NLRs bridge the gap to adaptive immunity by modulating and T-cell priming, for example through the regulation of (MHC) class I and II expression via NLRs like NLRC5 and CIITA. They also regulate key processes including , , and ; inflammasome-forming NLRs activate caspase-1 to promote , an inflammatory form of cell death that eliminates infected cells, while others like NLRX1 induce to clear intracellular threats. These functions collectively maintain immune balance, with NLRs integrating signals to fine-tune responses and prevent chronic activation. NLRs are widely expressed across cell types, including professional immune cells such as macrophages and dendritic cells, as well as non-immune tissues like epithelial barriers and neurons, allowing broad surveillance of potential threats. This ubiquitous expression underscores their significance in systemic immune and host protection. Dysregulation of NLR signaling, often through genetic variations, disrupts these balances and contributes to by promoting unchecked inflammation. Subfamily-specific roles, such as those of the NLRP and NLRC subfamilies in assembly, further highlight their diverse contributions to immunity.

Molecular Structure

Core Domains

NOD-like receptors (NLRs) exhibit a conserved that enables their role in innate immune sensing, consisting of an N-terminal effector domain, a central NACHT domain, and typically a C-terminal (LRR) domain. This modular structure, spanning approximately 800–1,200 in total depending on the receptor, allows NLRs to integrate detection with downstream signaling through protein oligomerization and interactions. The central NACHT domain, typically comprising 200–300 , serves as the core oligomerization module with an NTPase fold belonging to the AAA+ superfamily. This domain facilitates of NLR monomers into higher-order complexes upon activation by hydrolyzing like ATP, thereby acting as a for immune response initiation. Structurally, the NACHT domain includes a nucleotide-binding , helical domain 1, winged-helix domain, and helical domain 2, which together enable the conformational changes necessary for multimer formation. At the C-terminus, the LRR domain consists of tandem leucine-rich repeats, usually 200–300 in length, that provide specificity in recognizing diverse such as pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs). These repeats form a horseshoe-shaped structure that mediates ligand binding and autoregulation in the resting state, preventing premature activation. The N-terminal effector domain, varying in length but generally 50–150 , primarily functions in by recruiting downstream adaptor or effector proteins to propagate immune responses. It regulates NLR activity through homotypic interactions, enabling the assembly of signaling platforms while also contributing to autoinhibition in the inactive conformation. Subfamily-specific variations in this domain, such as different fold types, allow functional diversity across NLRs.

Domain Variations

NOD-like receptors (NLRs) display variations in their domain composition that distinguish subfamilies and underpin their diverse roles in innate immunity, while sharing a conserved tripartite architecture consisting of an N-terminal effector domain, a central NACHT oligomerization domain, and C-terminal leucine-rich repeats (LRRs). These effector domain differences occur primarily at the N-terminus, except in select cases like NLRP1, and determine subfamily classification into NLRA (acidic activation domain), NLRB (BIR domain), NLRC (CARD domain), and NLRP (PYD domain). The NLRC subfamily is characterized by an N-terminal caspase activation and recruitment domain (), which facilitates direct binding to kinases such as RIP2, as observed in NOD1 and NOD2. NLRC4 also contains this CARD domain, contributing to its distinct properties within the subfamily. The canonical structure for NLRC members, including NOD1 (also known as NLRC1) and (NLRC2), is CARD-NACHT-LRR, with the CARD positioned to mediate protein-protein interactions. In the NLRP subfamily, the N-terminal effector domain is a pyrin domain (PYD), which promotes interactions with adaptor proteins like ASC to support assembly, as exemplified by . The typical architecture is PYD-NACHT-LRR, with serving as a representative member due to its broad involvement in pathogen sensing. However, NLRP1 deviates from this pattern by incorporating a function-to-find (FIIND) C-terminal to the LRRs, followed by an additional , resulting in a PYD-NACHT-LRR-FIIND- structure. The FIIND in NLRP1 enables autoproteolytic cleavage between its ZU5 and UPA subdomains, a process critical for the protein's maturation and stability. Within the NLRP subfamily, NLRP10 is unique in lacking the LRR , resulting in a PYD-NACHT structure that may contribute to its distinct regulatory functions in immunity. The NLRB subfamily features N-terminal baculovirus protein repeat (BIR) domains, unique among NLRs for their role in apoptosis regulation and present in neuronal inhibitory proteins (NAIPs). NAIPs exhibit a BIR-NACHT-LRR architecture, with multiple BIR repeats (typically three) that distinguish them from other subfamilies and support specialized interactions. These domain variations across subfamilies allow NLRs to adapt their effector functions while maintaining LRR-mediated recognition. The NLRX subfamily, represented by NLRX1, exhibits a non-canonical structure lacking both a recognizable N-terminal effector (PYD, CARD, etc.) and LRRs; instead, it features a central NACHT flanked by unique N- and C-terminal regions that facilitate mitochondrial localization and antiviral signaling.

Classification and Nomenclature

Subfamily Categories

The NOD-like receptor (NLR) family in humans comprises 22 genes, categorized into five subfamilies based on the N-terminal effector architecture, with some members as pseudogenes or having limited functionality. This reflects structural similarities and shared evolutionary origins. The NLRA subfamily contains one member, CIITA (class II transactivator), featuring an acidic transactivating domain and involved in regulating expression. The NLRB subfamily, also known as NAIP (NLR family apoptosis inhibitory protein), includes one functional member in humans with baculoviral IAP repeat (BIR) domains; it senses bacterial effectors. Mice have seven NAIP paralogs (Naip1-7). The NLRC subfamily consists of five members (NLRC1 to NLRC5), characterized by a recruitment domain () and roles in activation and signaling. These include NLRC1 (also known as NOD1) and NLRC2 (), the first discovered NLRs involved in sensing; NLRC3 (also called NOD3), NLRC4 (also IPAF, detects bacterial and type III secretion systems), and NLRC5 (NOD4, regulates expression). The NLRP subfamily has 14 members (NLRP1 to NLRP14), defined by an N-terminal PYRIN (PYD) domain important for assembly. Examples include NLRP1 (involved in defenses, with unique C-terminal /FIIND) and (senses diverse DAMPs/PAMPs in ). Some, like NLRP10, lack leucine-rich repeats (LRRs) and have atypical or limited functions. The NLRX subfamily includes one member, NLRX1 (sometimes referred to as NOD5), with an unknown N-terminal and roles in mitochondrial antiviral signaling and regulation of . These five subfamilies account for all NLRs, highlighting their diversity in innate immune functions.

Evolutionary Relationships

NOD-like receptors (NLRs) represent an ancient family of intracellular sensors with origins tracing back to a common ancestor of and animals, where plant resistance (R) proteins serve as functional orthologs through shared nucleotide-binding and architectures essential for detection. These similarities in organization, including the central STAND (NACHT in animals, NB-ARC in ) and C-terminal LRRs, suggest of immune surveillance mechanisms across kingdoms, despite independent origins of the full NLR architecture in metazoans. NLR homologs are also present in fungi and protists, underscoring the broad phylogenetic distribution of this sensor class. In vertebrates, the NLR family underwent significant expansion through gene duplications, resulting in 22 functional NLR genes in humans, clustered into subfamilies NLRA, NLRB, NLRC, NLRP, and NLRX. This mammalian repertoire reflects ancient duplications predating eutherian divergence, with variations across species; for instance, fish exhibit diverse NLR expansions in some lineages but fewer canonical NACHT-containing members in others compared to mammals. Key evolutionary events include the segmental duplication of the NLRP cluster in following their divergence from approximately 90 million years ago, leading to multiple Nlrp4 paralogs in mice that are absent in humans. Conversely, experienced losses of certain NLR genes, such as those involved in detection, contributing to a more streamlined repertoire adapted to specific immune pressures. Sequence conservation within the NLR family is notably high in the central NACHT domain, which facilitates oligomerization and is a defining feature across vertebrates, while the LRR domains show greater variability to accommodate diverse ligand specificities. N-terminal effector domains, such as PYRIN or , exhibit even more divergence, reflecting subfamily-specific adaptations, though core sensor functions remain preserved from early metazoan ancestors. This pattern of conserved core elements amid variable peripheries highlights NLRs' evolutionary flexibility in innate immunity.

Activation Mechanisms

Ligand Recognition

NOD-like receptors (NLRs) detect microbial and endogenous danger signals through their C-terminal (LRR) domains, which serve as -sensing and autoregulatory regions in these intracellular pattern recognition receptors. In specific NLRs like NOD1 and , the LRR domains directly bind pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), while others such as respond indirectly to diverse signals that induce cellular perturbations. These LRR domains recognize a diverse array of PAMPs and DAMPs, initiating the innate immune response by distinguishing self from non-self or altered-self entities. Upon binding of a to the LRR domain, NLRs undergo a conformational rearrangement that disrupts intramolecular auto-inhibition, thereby exposing the central NACHT oligomerization domain. This structural shift allows the NACHT domain to become accessible for self-association, marking the critical transition from a resting to an active state without directly involving downstream signaling at this stage. The precision of this mechanism ensures that only relevant threats trigger NLR engagement, maintaining cellular . NLR ligands encompass both microbial and endogenous molecules, with representative examples including bacterial fragments as PAMPs that directly bind to LRR domains in select NLRs. Endogenous DAMPs, such as crystals released during cellular stress or injury, represent another major class, highlighting NLRs' role in sensing sterile inflammation. Ligand recognition can occur via direct interaction with the LRR, as seen with certain motifs, or indirectly through ligand-induced perturbations like efflux or lysosomal damage that propagate signals to the NLR. A key regulatory feature of NLR activation is the , which mandates integration of multiple signals to achieve full responsiveness and avert inappropriate immune activation leading to . Typically, this involves an initial priming signal—often from extracellular receptors like Toll-like receptors—that upregulates NLR expression or modifies their activity, followed by the specific PAMP or DAMP encounter to surpass the activation threshold. This multi-signal requirement fine-tunes NLR sensitivity, ensuring robust yet controlled responses to genuine threats.

Assembly Processes

Upon recognition, NOD-like receptors (NLRs) undergo conformational changes that initiate assembly into higher-order oligomeric complexes, primarily mediated by the central NACHT domain, which possesses nucleotide-binding and oligomerization capabilities. This domain, consisting of nucleotide-binding (NBD), helical domain 1 (), winged helix domain (WHD), and helical domain 2 (HD2) subdomains, facilitates ATP-dependent self-association, enabling the formation of wheel-like or disk-shaped structures essential for . For inflammasome-forming NLRs, oligomerization typically involves 7-11 subunits. For instance, forms an open octameric wheel-like structure via NACHT domain interactions, including back-to-back, head-to-face, tail-to-tail, and face-to-face contacts, as resolved by cryo-electron microscopy (cryo-EM); this assembly requires the co-factor NEK7, which binds the NACHT-LRR interface to license oligomerization. Similarly, NLRC4 assembles into disk-like oligomers of 10-11 subunits, nucleated by NAIP sensor proteins, with each subunit undergoing a ~90° hinge rotation at the NACHT domain to propagate the complex. Non-inflammasome NLRs like NOD1 and form distinct oligomeric platforms, often filamentous structures with adaptor proteins. These structures provide a scaffold for downstream interactions, with the number of subunits influencing the stability and efficiency of assembly. Adaptor recruitment follows oligomerization and varies by NLR architecture. NLRPs, which contain a pyrin domain (PYD), recruit the adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC) through homotypic PYD-PYD interactions; ASC then polymerizes into filaments and binds pro-caspase-1 via CARD-CARD contacts. In contrast, CARD-containing NLRs like NOD1 and directly recruit receptor-interacting protein kinase 2 (RIP2) or caspase-1 without ASC, forming signaling platforms for activation. Assembly distinguishes inflammasome-forming NLRs from non-inflammasome ones. , such as those involving , NLRP1, or NLRC4, culminate in visible ASC specks—dense, punctate aggregates observable by —that amplify caspase-1 activation and promote . Non-inflammasome NLRs, like NOD1 and , form transient oligomers without speck formation, instead prioritizing adaptor-mediated signaling. This speck-based assembly in enhances and signal amplification within the . Regulation of assembly is tightly controlled to prevent aberrant activation. ATP binding and hydrolysis by the NACHT domain are indispensable, driving nucleotide-dependent oligomerization while inhibitors like MCC950 disrupt this process in NLRP3. Post-translational modifications further modulate dynamics; for example, K63-linked ubiquitination stabilizes NLRP3 oligomers, whereas K48-linked ubiquitination targets them for proteasomal degradation, with deubiquitinases like BRCC3 promoting activation. Phosphorylation and other modifications, such as S-nitrosylation, also fine-tune assembly thresholds.

Signaling Pathways

NOD Subfamily Pathways

NOD1 and NOD2, the primary members of the NOD subfamily, are intracellular pattern recognition receptors that detect specific bacterial peptidoglycan-derived motifs, initiating signaling cascades critical for innate immune responses. NOD1 recognizes γ-D-glutamyl-meso-diaminopimelic acid (iE-DAP or GM-tripeptide), a component prevalent in Gram-negative and certain Gram-positive bacteria, while NOD2 senses muramyl dipeptide (MDP), a motif found in peptidoglycan from both Gram-positive and Gram-negative bacteria. Upon ligand binding to their leucine-rich repeat domains, both receptors undergo conformational changes that promote oligomerization and expose their caspase activation and recruitment domains (CARDs). These CARD domains then facilitate homotypic interactions with the CARD of the adaptor protein receptor-interacting serine/threonine-protein kinase 2 (RIP2, also known as RIPK2 or RICK), recruiting it to the receptor complex and initiating downstream signaling. The core signaling pathway downstream of NOD1 and NOD2 activation centers on RIP2-mediated ubiquitination events that drive pro-inflammatory responses. Recruited RIP2 undergoes lysine-63 (K63)-linked polyubiquitination, primarily at lysine 209 in its kinase domain, which is essential for signal propagation and independent of RIP2's kinase activity. This ubiquitination recruits the TAB1/TAB2/TAK1 complex, where transforming growth factor-β-activated kinase 1 (TAK1) phosphorylates the IκB kinase (IKK) complex, including IKKα, IKKβ, and NEMO. Activated IKK then phosphorylates the inhibitory protein IκBα, leading to its K48-linked ubiquitination, proteasomal degradation, and subsequent nuclear translocation of NF-κB dimers. In the nucleus, NF-κB binds to promoter regions of target genes, inducing expression of pro-inflammatory cytokines such as IL-6, IL-8, and TNF-α, as well as antimicrobial peptides. Additionally, the pathway activates mitogen-activated protein kinases (MAPKs), including p38 and JNK, through TAK1-mediated phosphorylation, contributing to further amplification of inflammatory gene expression and cellular responses like apoptosis in certain contexts. Beyond and MAPK signaling, NOD1 and promote as an alternative output to enhance bacterial clearance. Upon detecting invasive , NOD1 and independently of RIP2 recruit autophagy-related protein 16-like 1 (ATG16L1) to the plasma membrane at sites of bacterial entry via direct interaction with bacterial . This localization facilitates formation around intracellular pathogens, targeting them for lysosomal degradation and limiting infection. Dysregulation of this process, as seen in Crohn's disease-associated variants, impairs autophagic bacterial handling. Signaling through the NOD-RIP2 axis is tightly regulated to prevent excessive inflammation, primarily through ubiquitin-modifying enzymes that provide . The E3 promotes K63-linked ubiquitination of RIP2, which facilitates its proteasomal degradation and attenuates sustained and MAPK activation following NOD stimulation. Additionally, S-palmitoylation of and by ZDHHC5 facilitates their membrane recruitment, enhancing signaling efficiency. Similarly, the deubiquitinase CYLD removes both K63- and linear (Met1) chains from RIP2, limiting its oligomerization and downstream TAK1/IKK signaling to dampen pro-inflammatory outputs. These regulatory mechanisms ensure balanced immune responses, with deficiencies in or CYLD linked to hyperinflammatory states.

NLRP Subfamily Pathways

The NLRP subfamily of NOD-like receptors (NLRs) primarily functions through the formation of , multiprotein complexes that drive caspase-1 activation and subsequent inflammatory responses. Key members, including NLRP1, (also known as cryopyrin), and NLRP6, assemble ASC-dependent upon sensing diverse danger signals or pathogens. These proteins share a typical NLR with a pyrin domain (PYD) at the , a central NACHT oligomerization domain, and C-terminal leucine-rich repeats (LRRs) for sensing, though NLRP1 uniquely features a function-to-find domain (FIIND) and . Upon activation, NLRP oligomerization facilitates recruitment of the adaptor protein apoptosis-associated speck-like protein containing a (ASC) via PYD-PYD interactions, followed by ASC's domain binding pro-caspase-1 to induce its dimerization and autocleavage into active caspase-1. Active caspase-1 then processes pro-interleukin-1β (IL-1β) and pro-IL-18 into their mature, secreted forms, while cleaving gasdermin D (GSDMD) to form N-terminal pores that execute , an inflammatory lytic . NLRP3 stands out as a versatile multi-ligand activated by a broad array of stimuli, including extracellular ATP (via P2X7 receptor) and bacterial ionophores like nigericin, which disrupt cellular . Its activation follows a two-signal model: the priming signal (Signal 1), often from ligands like (LPS), induces NF-κB-dependent transcriptional upregulation of NLRP3 and pro-IL-1β. The activation signal (Signal 2) then triggers NLRP3 oligomerization through integrated stress responses, such as (K⁺) efflux, mitochondrial (ROS) production, and lysosomal membrane damage releasing cathepsins. These upstream events converge to relieve autoinhibition, enabling ASC speck formation and caspase-1 activation, with NEK7 kinase serving as a critical co-factor for NLRP3-ASC interaction. Additionally, NLRP3 activation involves deubiquitination by BRCC3 and ISGylation, while such as β-hydroxybutyrate suppress its activity to limit . In contrast, NLRP1 activation involves a distinct proteolytic mechanism, where pathogen-derived proteases (e.g., from Bacillus anthracis lethal factor or viral 3C-like proteases) or cellular stresses cleave NLRP1 at specific sites, leading to proteasome-mediated degradation of its N-terminal inhibitory fragment and release of the FIIND-CARD fragment for auto-processing and ASC recruitment. NLRP6, while less extensively characterized, forms ASC- and caspase-1-dependent inflammasomes in response to microbial products like lipoteichoic acid (LTA) and double-stranded RNA, following a similar two-step priming and activation process that promotes IL-1β/IL-18 release and pyroptosis, particularly in intestinal epithelial cells. Regulation across the NLRP subfamily involves post-translational modifications, such as deubiquitination for NLRP3, and inhibitors like DPP8/9 for NLRP1, ensuring tightly controlled inflammasome responses to prevent excessive inflammation.

NLRC and NAIP Subfamily Pathways

The NAIP-NLRC4 represents a key signaling pathway in the NLRC and NAIP subfamilies, specialized for detecting bacterial components from Gram-negative pathogens. NAIP proteins, particularly NAIP2 and NAIP5 in mice (with human homologs NAIP functioning similarly), directly recognize specific bacterial motifs such as (via NAIP5/NAIP2) or (T3SS) needle and rod proteins (via NAIP1/NAIP2). Upon ligand binding, NAIPs undergo conformational changes that expose their nucleotide-binding (NACHT) and C-terminal domains, enabling direct CARD-CARD interactions with NLRC4 (also known as IPAF). This interaction initiates a self-propagating oligomerization process, forming a wheel-like complex that recruits the adaptor protein ASC (apoptosis-associated speck-like protein containing a ) through additional CARD interactions. The assembled NAIP-NLRC4 inflammasome activates caspase-1 via proximity-induced autocleavage, leading to the processing and release of pro-inflammatory cytokines IL-1β and IL-18, as well as gasdermin D-mediated in infected cells. Unlike some other , NAIP-NLRC4 activation can occur independently of priming in certain contexts, allowing rapid responses to cytosolic bacterial invasion without prior transcriptional upregulation. This pathway is particularly effective against like Salmonella, , and , where T3SS components serve as potent activators, highlighting its role in restricting intracellular pathogen replication. In contrast to the inflammasome-forming members, NLRC5 in the NLRC subfamily functions primarily as a transcriptional rather than a sensor-adaptor. NLRC5 transactivates genes (HLA-A, -B, -C in humans) by binding to the SXY module in their promoters and recruiting histone acetyltransferases like RFXANK and , independent of signaling. This regulation enhances to CD8+ T cells, crucial for antiviral and antitumor immunity, and occurs through NLRC5's nuclear translocation and interaction with the enhanceosome complex. NLRC5 expression is induced by interferons, amplifying surface expression without direct involvement in assembly. Beyond transcriptional regulation, NLRC5 associates with NLRP12 in the NLRC5-PANoptosome to trigger PANoptosis, releasing cytokines and DAMPs, regulated by TLR2/TLR4 and NAD+.

NLRX Subfamily Pathways

NLRX1, the sole member of the NLRX subfamily, localizes to mitochondria and negatively regulates NF-κB and type I interferon (IFN) signaling to maintain immune homeostasis. It interacts with mitochondrial proteins like MAVS (mitochondrial antiviral-signaling protein) to dampen excessive antiviral responses and modulates reactive oxygen species (ROS) production. Upon sensing viral double-stranded DNA (e.g., from HSV-1) or bacterial components via its LRR domain, NLRX1 enhances its ATPase activity and regulates the STING-TBK1 pathway, fine-tuning type I IFN production and preventing hyperinflammation. NLRX1 also promotes autophagy and bacterial clearance in the gut, with dysregulation linked to inflammatory bowel disease and viral susceptibility.

Physiological Functions

Role in Innate Immunity

NOD-like receptors (NLRs) serve as critical intracellular sensors in the , detecting microbial motifs and danger signals to orchestrate pathogen clearance. In defense against bacteria, NOD2 promotes xenophagy, a selective form of that targets intracellular pathogens for lysosomal degradation, thereby enhancing bacterial killing in epithelial cells and dendritic cells. Similarly, NLRP3 and NLRC4 activate that trigger , a lytic form of in macrophages, which releases engulfed bacteria for subsequent neutrophil-mediated destruction and limits intracellular replication of pathogens such as Salmonella typhimurium and . These mechanisms ensure rapid elimination of invaders without relying solely on signaling. NLRs coordinate innate responses by integrating with other pattern recognition receptors and bridging to adaptive immunity. Crosstalk between NLRs and Toll-like receptors (TLRs) amplifies inflammation; for instance, TLR-induced NF-κB activation produces pro-IL-1β, which NLR inflammasomes then process into mature IL-1β via caspase-1, heightening antimicrobial activity. Additionally, NLRC5 functions as a transcriptional transactivator for MHC class I genes, upregulating expression of HLA-A, -B, -C, and associated components like TAP1 and LMP2 to enhance antigen presentation to CD8⁺ T cells, thereby linking innate detection to cytotoxic responses against infected cells. In tissue-specific contexts, NLRs tailor immunity to local threats while preserving barrier functions. In the gut , NOD2 recognizes muramyl dipeptide from commensal and , maintaining epithelial integrity by promoting production of and supporting survival, which reinforces the mucosal barrier against translocation. In the lungs, NLRP3 in alveolar macrophages senses viral and danger signals during infection, driving IL-1β release to recruit neutrophils and promote viral clearance without excessive tissue damage. NLRs also uphold intestinal by establishing tuned activation thresholds that foster tolerance to commensal . Through regulated sensing of microbial , NLRs like NOD1, , and NLRP6 modulate signaling and activity to prevent , support epithelial repair, and limit unnecessary , ensuring a balanced coexistence with the gut flora. This selective responsiveness distinguishes harmless residents from threats, sustaining immune quiescence in steady-state conditions.

Involvement in Autoinflammation

NOD-like receptors (NLRs) contribute to autoinflammation through dysregulated activation that promotes sterile inflammatory responses in the absence of infection. Gain-of-function mutations in lead to constitutive assembly and excessive interleukin-1β (IL-1β) production, driving systemic such as cryopyrin-associated periodic syndromes (CAPS). These mutations, often located in the nucleotide-binding domain of , lower the activation threshold, enabling spontaneous oligomerization with ASC and caspase-1 without typical danger signals, resulting in chronic low-grade inflammation characterized by fever, rash, and joint pain. Defects in impair autophagy pathways, exacerbating autoinflammatory conditions by failing to clear damaged cellular components and maintain . Loss-of-function mutations in , common in , disrupt its interaction with ATG16L1, hindering autophagosome formation and leading to accumulation of ubiquitinated aggregates that trigger hyperactivation and persistent release. This autophagy deficiency promotes unresolved in the intestinal mucosa, contributing to granulomatous tissue damage and chronic autoinflammatory flares. In metabolic disorders, NLRP3 senses endogenous danger signals like crystals, linking NLR activation to sterile in . crystals from ruptured plaques are phagocytosed by macrophages, inducing lysosomal damage and efflux that prime and activate the , amplifying IL-1β and IL-18 secretion to drive plaque instability and vascular . This mechanism underscores NLRP3's role in amplifying metabolic stress into chronic inflammatory cascades. Certain NLRs, such as NLRP12, act as negative regulators to resolve autoinflammation by suppressing excessive signaling. NLRP12 inhibits and MAPK pathways in response to self-ligands, preventing overproduction of proinflammatory cytokines and maintaining immune in tissues like the colon. Deficiency in NLRP12 leads to heightened susceptibility to autoinflammatory phenotypes, highlighting its essential role in dampening sterile inflammatory responses.

Disease Associations

Genetic Disorders

Mutations in genes encoding NOD-like receptors (NLRs) underlie several monogenic autoinflammatory disorders characterized by dysregulated innate immune responses. These conditions arise primarily from gain-of-function variants that lead to excessive activation and production, contrasting with loss-of-function variants that confer susceptibility to complex diseases. patterns typically follow autosomal dominant transmission for gain-of-function , while susceptibility alleles exhibit more complex, often polygenic . Blau syndrome and early-onset sarcoidosis represent autoinflammatory granulomatous diseases caused by missense mutations in the gene (also known as CARD15) that disrupt its regulatory functions, leading to dysregulated signaling and unchecked inflammation. manifests in early childhood with a triad of granulomatous , symmetric , and , progressing to potential vision loss and joint deformities. It is inherited in an autosomal dominant manner due to heterozygous missense mutations, predominantly in the nucleotide-binding domain of . Early-onset sarcoidosis shares identical clinical features but occurs sporadically, often from mutations in the same gene, confirming a common genetic with . In contrast, loss-of-function variants, such as frameshift or nonsense mutations (e.g., L1007fs), impair bacterial sensing and are associated with increased susceptibility to , a multifactorial inflammatory bowel disorder, through reduced and impaired mucosal barrier function. Gain-of-function mutations in NLRP1 are linked to rare autoinflammatory skin disorders, including NLRP1-associated autoinflammation with and dyskeratosis (NAIAD). This features chronic skin inflammation with dyskeratosis, , and elevated levels of caspase-1 and IL-18, reflecting hyperactive NLRP1 assembly. Inheritance can be autosomal recessive, as seen in homozygous missense variants (e.g., p.Arg726Trp), or sporadic via de novo heterozygous mutations (e.g., p.Pro1214Arg). NLRP1 polymorphisms also confer susceptibility to , an autoimmune skin depigmentation disorder, by promoting inflammasome-mediated keratinocyte death and autoantigen exposure. Cryopyrin-associated periodic syndromes (CAPS) encompass a spectrum of autoinflammatory conditions driven by gain-of-function mutations in the gene, encoding cryopyrin. These heterozygous variants, often in the NACHT domain, cause constitutive activation and overproduction of IL-1β, leading to episodic fever, rash, and organ-specific inflammation. The mildest form, familial cold autoinflammatory syndrome (FCAS), presents with cold-triggered urticaria and . Muckle-Wells syndrome involves recurrent urticaria, sensorineural deafness, and risk. The severe neonatal-onset multisystem inflammatory disease (NOMID) includes chronic , , and growth failure. All CAPS subtypes follow autosomal dominant , with early IL-1 therapy preventing long-term complications.

Infectious and Chronic Diseases

NOD-like receptors (NLRs) play a critical role in host defense against bacterial pathogens, with dysfunction increasing susceptibility to infections. Variants in , a key NLR involved in sensing muramyl dipeptide from bacterial , have been associated with heightened risk of infections by intracellular bacteria such as and . In NOD2-deficient macrophages, impaired activation and production, including IL-12 and TNF-α, lead to reduced bacterial clearance and exacerbated disease progression in experimental models. Similarly, NOD2 polymorphisms in humans correlate with increased vulnerability to mycobacterial infections, highlighting its importance in innate immune responses at mucosal barriers. Hyperactivation of NLRC4, another NLR that detects bacterial via NAIP proteins, contributes to autoinflammatory during infections. Gain-of-function mutations in NLRC4 cause excessive assembly, leading to overproduction of IL-1β and IL-18, which drive intestinal inflammation and in response to enteric pathogens. This manifests as severe, recurrent with diarrhea and tissue damage, as observed in patients with NLRC4-associated autoinflammatory disease (AID). In mouse models, NLRC4 hyperactivation in intestinal epithelial cells promotes cell expulsion and release, exacerbating pathology during bacterial challenges like . In chronic diseases, NLR dysregulation links to metabolic and inflammatory conditions through persistent activation. NLRP3 senses urate crystals in , triggering caspase-1-mediated IL-1β release and acute ; this pathway is central to monosodium urate (MSU)-induced flares, as demonstrated in early studies showing knockout prevents MSU-driven in mice. High glucose levels in similarly activate via ROS accumulation and ATP-P2X4 signaling, promoting , , and β-cell , which worsen glycemic control and vascular complications. Some studies suggest NOD2 variants contribute to (RA) pathogenesis in certain populations, with polymorphisms such as rs3135500 associating with elevated RA risk and disease activity. NLRP3 exhibits dual roles in cancer, promoting tumorigenesis through IL-1β-driven and immune suppression while offering protection via in some contexts. Chronic NLRP3 activation in tumor-associated macrophages fosters a pro-tumor microenvironment by enhancing IL-1β secretion, which supports tumor growth and metastasis in models of and . Conversely, NLRP3-induced eliminates infected or transformed cells, limiting viral oncogenesis and early tumor formation, as seen in where NLRP3 deficiency accelerates progression. This ambivalence underscores NLRP3 as a therapeutic target in . Therapeutic strategies targeting NLRs, particularly NLRP3 inhibitors, show promise for managing infectious and chronic diseases as of 2025. MCC950, a potent antagonist, demonstrated efficacy in preclinical models of and by blocking IL-1β release, though its clinical development was halted due to . Next-generation inhibitors, including derivatives, are advancing in phase I/II trials for autoinflammatory conditions with potential extensions to chronic inflammatory diseases; for example, VTX958 from Ventyx Biosciences is in phase II trials for and , showing promising safety and efficacy profiles as of 2025. These agents inhibit assembly without off-target effects, offering hope for reducing infection susceptibility and ameliorating RA and metabolic syndromes.

References

  1. [1]
  2. [2]
    NOD-Like Receptors: Versatile Cytosolic Sentinels
    NLRs are expressed intracellularly and have been shown to respond to a wide variety of classes of ligands, bacterial wall components, toxins, and host-derived ...Skip main navigation · Abstract · Invertebrates · Inflammasome Assembly
  3. [3]
    Unleashing the therapeutic potential of NOD-like receptors - Nature
    NLRs trigger innate immune responses by inducing signalling pathways, such nuclear factor-κB, mitogen-activated protein kinases, and the caspase 1 inflammasome.
  4. [4]
    Function of Nod-like Receptors in Microbial Recognition and Host ...
    This review focuses on NLRs, which are primarily involved in bacterial recognition. An important finding has been the discovery that immune dysregulation ...
  5. [5]
    Nod1, an Apaf-1-like activator of caspase-9 and nuclear factor-kappaB
    May 21, 1999 · Nod1 is a leucine-rich repeat-containing Apaf-1-like molecule that can regulate both apoptosis and NF-kappaB activation pathways.
  6. [6]
    An induced proximity model for NF-kappa B activation in the Nod1 ...
    We show that self-association of Nod1 mediates proximity of RICK and the interaction of RICK with the gamma subunit of the IkappaB kinase (IKKgamma).
  7. [7]
    Association of NOD2 leucine-rich repeat variants with susceptibility ...
    These observations suggest that the NOD2 gene product confers susceptibility to Crohn's disease by altering the recognition of these components.
  8. [8]
    Plant NBS-LRR proteins: adaptable guards - PMC - PubMed Central
    Plant NBS-LRR proteins are similar in sequence to members of the mammalian nucleotide-binding oligomerization domain (NOD)-LRR protein family (also called ' ...
  9. [9]
    Defining the Origins of the NOD-Like Receptor System at the Base of ...
    In vertebrates, two major types of NLR complexes can be defined: 1) in the NODosome (NOD1/NOD2), the serine–threonine kinase receptor-interacting serine/ ...Introduction · Materials and Methods · Results · Discussion
  10. [10]
    NOD-Like Receptors: Master Regulators of Inflammation and Cancer
    Cytosolic NOD-like receptors (NLRs) have been associated with human diseases including infections, cancer, and autoimmune and inflammatory disorders.
  11. [11]
    The role of NOD-like receptors in innate immunity - PMC
    Mar 15, 2023 · The NLRs play a key role in sensing molecules associated with intracellular infection and stress. Thus, they sense a variety of generic stimuli ...
  12. [12]
    The role of NOD-like Receptors in shaping adaptive immunity - PMC
    A diverse class of PRRs, the NOD-like Receptors (NLR), has recently been implicated in the regulation of processes ranging from anti-tumor immunity to the ...Prrs In Innate... · Table 1 · Inflammasome-Adaptive...
  13. [13]
  14. [14]
    The role of NOD-like receptors in innate immunity - Frontiers
    Mar 14, 2023 · Overall, NOD1 and NOD2 signaling is important during host defense against microbes, and is associated with metabolic, autoimmune, and ...
  15. [15]
    NOD-Like Receptors in Infection, Immunity, and Diseases - PMC
    There are four recognizable N-terminal domains, which are used to classify NLRs into four subfamilies: the acidic transactivation domain (NLRA), the baculoviral ...
  16. [16]
  17. [17]
  18. [18]
    The NLRP1 inflammasome: new mechanistic insights and ...
    May 20, 2019 · In NLRP1 and CARD8, the FIIND is followed by a C-terminal CARD, a death-fold domain. This domain architecture is found in several other human ...
  19. [19]
    Autolytic Proteolysis within the Function to Find Domain (FIIND) Is ...
    Here, we identify the requirements for activation of NLRP1, an NLR protein associated with a number of human pathologies, including vitiligo, rheumatoid ...
  20. [20]
    The NLR gene family: from discovery to present day - PubMed Central
    The mammalian nucleotide-binding domain leucine-rich repeat containing (NLR, NBD-LRR or NOD-like receptor) family was reported over 20 years ago.
  21. [21]
    Human NAIP and mouse NAIP1 recognize bacterial type III secretion ...
    Aug 12, 2013 · NAIPs are a family of NLRs with seven paralogs in mice (NAIP1–7) but only one family member in humans (hNAIP). NAIP5/6 and NAIP2 bind directly ...
  22. [22]
    Animal NLRs provide structural insights into plant NLR function - PMC
    Plant NLRs show structural and functional resemblance to animal NLRs involved in inflammatory and innate immune responses.
  23. [23]
    The NBS-LRR architectures of plant R-proteins and metazoan NLRs ...
    Jan 17, 2017 · We find that the NBS-LRR domain architecture of R-proteins and NLR proteins likely evolved at least twice, independently in plants and metazoans ...
  24. [24]
    Plant NLR diversity: the known unknowns of pan-NLRomes - PMC
    NLR proteins are found in plants, animals, fungi, and protists, although the similarities in protein architecture are thought to result from convergent ...
  25. [25]
    A genomic view of the NOD-like receptor family in teleost fish
    Feb 6, 2008 · Three distinct NLR subfamilies were identified by mining genome databases of various non-mammalian vertebrates; the first subfamily (NLR-A) ...
  26. [26]
    Evolution and functional divergence of NLRP genes in mammalian ...
    We found that (1) major NLRP genes have been duplicated before the divergence of mammals, with certain lineage-specific duplications in primates (NLRP7 and 11) ...
  27. [27]
    NOD2 and reproduction-associated NOD-like receptors have been ...
    Nov 1, 2021 · We conclude that parallel evolution has led to the loss of both NLR proteins involved in intracellular flagellin detection in pangolins and ...
  28. [28]
    The Nod-Like Receptor (NLR) Family: A Tale of Similarities and ...
    NLRs are believed to initiate or regulate host defense pathways through formation of signaling platforms that subsequently trigger the activation of ...
  29. [29]
    Pattern recognition receptors in health and diseases - Nature
    Aug 4, 2021 · PRRs are basically composed of ligand recognition domains, intermediate domains, and effector domains. PRRs recognize and bind their respective ...
  30. [30]
    Nod‐like receptors in innate immunity and inflammatory diseases
    Jul 8, 2009 · Inohara N., Chamaillard M., McDonald C., Nunez G. Nod‐LRR proteins: role in host‐microbial interactions and inflammatory disease. Annu Rev ...
  31. [31]
    Gout-associated uric acid crystals activate the NALP3 inflammasome
    Jan 11, 2006 · Martinon, F., Pétrilli, V., Mayor, A. et al. Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature 440, 237–241 (2006).
  32. [32]
    Cryo-EM structure of the activated NAIP2-NLRC4 inflammasome ...
    Oct 8, 2015 · We found that the PrgJ-NAIP2-NLRC4 inflammasome is assembled into multisubunit disk-like structures through a unidirectional adenosine triphosphatase ...
  33. [33]
    Inflammasome assembly: The wheels are turning | Cell Research
    Nov 24, 2015 · ... wheel-like structure and thus facilitates progressive oligomerization (Figure 1). ... Cryo-EM structure of such complexes has not yet been solved.
  34. [34]
  35. [35]
    Structural basis for distinct inflammasome complex assembly by ...
    Jan 8, 2021 · NLRP1 and CARD8 both harbor a poorly defined auto-proteolytic domain known as FIIND, whose auto-cleavage is required for inflammasome activation ...
  36. [36]
  37. [37]
    A critical role of RICK/RIP2 polyubiquitination in Nod-induced NF-κB ...
    The induction of immune response genes through Nod1 and Nod2 signaling largely depends on NF-κB, a proinflammatory transcriptional factor (Inohara et al, 1999, ...Missing: seminal papers
  38. [38]
    Roles of NOD1 (NLRC1) and NOD2 (NLRC2) in innate immunity ...
    Gene KO (knockout) studies indicate that RIP2 is a critical mediator of NOD1 and NOD2 signallings [34–36], though precise details are unclear. Major NOD- ...<|control11|><|separator|>
  39. [39]
    Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to ... - Nature
    Nov 8, 2009 · Here we demonstrated that the intracellular sensors Nod1 and Nod2 are critical for the autophagic response to invasive bacteria.
  40. [40]
    Article CYLD Limits Lys63- and Met1-Linked Ubiquitin at Receptor ...
    Mar 29, 2016 · This prompted us to explore the role of CYLD in regulating the NOD2 pathway, which relies on LUBAC. CYLD Activity Controls NOD2 Signaling. CYLD ...
  41. [41]
    NOD Signaling and Cell Death - Frontiers
    To better understand the connections between NOD signaling and cell death induction, we here review the latest findings on the molecular regulation of signaling ...
  42. [42]
    NLRP inflammasomes in health and disease | Molecular Biomedicine
    Apr 22, 2024 · Here, we provide the latest research progress on NLRP inflammasomes, including NLRP1, CARD8, NLRP3, NLRP6, NLRP7, NLRP2, NLRP9, NLRP10, and ...
  43. [43]
    Mechanism of NLRP3 inflammasome activation - PMC
    Considering the potency of the products of inflammasome activation, the requirement for two signals to achieve activation represents an important regulatory ...
  44. [44]
    Mechanism and regulation of NLRP3 inflammasome activation - PMC
    In this review we summarize current understanding of the mechanism and regulation of NLRP3 inflammasome activation, as well as recent advances.
  45. [45]
    The NLRP3 Inflammasome Pathway: A Review of Mechanisms and ...
    Upon activation by PAMPs and DAMPs, NLRP3 oligomerizes and activates caspase-1 which initiates the processing and release of pro-inflammatory cytokines IL-1β ...
  46. [46]
    Updated insights into the molecular networks for NLRP3 ... - Nature
    Apr 30, 2025 · In this Review, we provide the current understanding of the complex molecular networks that play pivotal roles in regulating NLRP3 inflammasome priming, ...
  47. [47]
    Structural Mechanisms of NLRP3 Inflammasome Assembly and ...
    Apr 26, 2023 · This article discusses the structural mechanisms of NLRP3 inflammasome assembly and activation. Authors are Jianing Fu and Hao Wu.
  48. [48]
    Diverse viral proteases activate the NLRP1 inflammasome - eLife
    Jan 7, 2021 · NLRP1 has an unusual domain architecture, containing a CARD at its C-terminus rather than the N-terminus like all other inflammasome sensor NLRs ...
  49. [49]
    The NLRP6 inflammasome - PMC - PubMed Central
    In this review, we summarize recent understandings on the assembly, activation and regulation patterns of the NLRP6 inflammasome, as well as its diverse ...
  50. [50]
    Mechanism of NAIP—NLRC4 inflammasome activation revealed by ...
    Jan 5, 2023 · After activation, NAIPs can activate NLRC4 and form the wheel-like NAIP—NLRC4 inflammasome complex, which further recruits and activates the ...
  51. [51]
    Structural basis of the human NAIP/NLRC4 inflammasome assembly ...
    Jan 4, 2024 · Especially in humans, the NLR family apoptosis inhibitory protein (NAIP)/NLRC4 inflammasome has been shown to be triggered after the sensing of ...Missing: NLRB members
  52. [52]
    Emerging Concepts about NAIP/NLRC4 Inflammasomes - PMC
    Specific NAIP proteins bind to the agonists and then physically associate with NLRC4 to form an inflammasome complex able to recruit and activate pro-caspase-1.Missing: seminal papers
  53. [53]
    NLRC4 biology in immunity and inflammation - Andrade - 2020
    Jun 12, 2020 · In this review, we discuss recent advances on the biology of the NLRC4 inflammasome, in terms of structure and activation mechanisms, importance in bacterial ...
  54. [54]
    NLR family member NLRC5 is a transcriptional regulator of ... - PNAS
    Our results suggest that NLRC5 is a transcriptional regulator, orchestrating the concerted expression of critical components in the MHC class I pathway.
  55. [55]
    NLRC5 regulates MHC class I antigen presentation in host defense ...
    Apr 10, 2012 · NLRC5 is a critical regulator of host defense against intracellular pathogens in vivo. NLRC5 was specifically required for the expression of genes involved in ...
  56. [56]
    MHC class I transactivator NLRC5 in host immunity, cancer and ...
    NLRC5 is a transcriptional co‐activator of MHC class I and related genes. NLRC5 plays critical roles in defense against viral infection and cancer.Missing: seminal | Show results with:seminal
  57. [57]
    Page not found | Nature
    Insufficient relevant content.
  58. [58]
  59. [59]
  60. [60]
    Review Toll-like receptors (TLRs) and Nod-like receptors (NLRs) in ...
    More recently, accumulating evidence suggests that NLRs complement and synergize with TLRs in induction of innate immune responses. Various levels of crosstalk ...Review · Introduction · Roles Of Tlrs And Nlrs In...
  61. [61]
    Recognition of gut microbiota by NOD2 is essential for the ...
    NOD2 signaling maintains intestinal intraepithelial lymphocytes via recognition of gut microbiota and IL-15 production.
  62. [62]
    NLRP3 Inflammasome—A Key Player in Antiviral Responses - PMC
    Feb 18, 2020 · The NLRP3 inflammasome is the most extensively studied and it plays an important role in both inflammation and antiviral responses.
  63. [63]
    NOD-Like Receptors in Intestinal Homeostasis and Epithelial Tissue ...
    NOD-Like Receptors (NLRs) in Intestinal Homeostasis, Inflammation and Tissue Repair. The human NOD-like receptor (NLR) family of intracellular sensors comprises ...Nod-Like Receptors In... · 5. Innate Immunity... · 5.2. 2. Inflammasome Nlrs<|control11|><|separator|>
  64. [64]
    CAPS and NLRP3 - PMC - PubMed Central - NIH
    CAPS knockin mutant mouse models were generated with Nlrp3 mutations observed in FCAS, MWS, and CINCA/NOMID patients to further investigate mechanisms involved ...
  65. [65]
    Crohn's disease: NOD2, autophagy and ER stress converge - PMC
    Jan 19, 2011 · Polymorphisms in NOD2, encoding an intracellular pattern recognition receptor, contribute the largest fraction of genetic risk for Crohn's disease.
  66. [66]
    Cholesterol Crystals Activate the NLRP3 Inflammasome in Human ...
    Our study shows that the same activation pathway of NLRP3 is functional also in human macrophages exposed to cholesterol crystals. Furthermore, Duewell and ...
  67. [67]
    NLRP12 in Innate Immunity and Inflammation - PMC - PubMed Central
    (A) NLRP12 negatively regulates inflammatory signaling by suppressing canonical and non-canonical NFκB signaling and the MAPK/ERK signaling pathway in bone ...
  68. [68]
    Nucleotide-Binding Oligomerization Domain Protein 2-Deficient ...
    NOD2 has recently been shown to be important for host cell cytokine responses to Mycobacterium tuberculosis, to synergize with Toll-like receptor 2 (TLR2) in ...
  69. [69]
    NOD2, an intracellular innate immune sensor involved in host ...
    Jul 13, 2011 · These studies together establish that NOD proteins participate in the induction of autophagy and, as such, have a role in both antigen ...Rick-Dependent Nod2... · Nod2 Participation In Innate... · Nod2 And Crohn's Disease
  70. [70]
    A role for membrane-bound CD147 in NOD2-mediated recognition ...
    Feb 15, 2008 · Functional studies have shown that NOD2 in humans may be important for the recognition of a diverse range of pathogens including Listeria spp., ...Cd147/emmprin Interacts With... · Cd147 Negatively Regulates... · Cd147 Expression Enhances...
  71. [71]
    Mutation of NLRC4 causes a syndrome of enterocolitis and ...
    An activating NLRC4 inflammasome mutation causes a novel autoinflammatory syndrome presenting with recurrent Macrophage Activation Syndrome. Nature Genetics ...
  72. [72]
    Recessive NLRC4-Autoinflammatory Disease Reveals an Ulcerative ...
    Nov 16, 2021 · NLRC4-associated autoinflammatory disease (NLRC4-AID) is an autosomal dominant condition presenting with a range of clinical manifestations ...
  73. [73]
    Gout-associated uric acid crystals activate the NALP3 inflammasome
    Mar 9, 2006 · Here we show that MSU and CPPD engage the caspase-1-activating NALP3 (also called cryopyrin) inflammasome, resulting in the production of active interleukin ( ...Missing: paper | Show results with:paper
  74. [74]
    induced endothelial dysfunction and NLRP3 inflammasome activation
    Aug 15, 2019 · Activation of the NLRP3 inflammasome plays an important role in high glucose- induced endothelial dysfunction in patients with type 2 diabetes
  75. [75]
    ATP-P2X4 signaling mediates NLRP3 inflammasome activation
    Feb 19, 2013 · ATP-P2X4 signaling mediates high glucose-induced activation of the NLRP3 inflammasome, regulates IL-1 family cytokine secretion, and causes the development of ...
  76. [76]
    Association of rs3135500 and rs3135499 Polymorphisms in the ...
    Our findings propose a considerable association between NOD2 polymorphisms with increased risk of RA and disease activity.
  77. [77]
    NLRP3 Inflammasome Activation in Cancer: A Double-Edged Sword
    Jul 8, 2020 · The formation of inflammasome complexes leads to the activation of caspase-1, production of interleukin (IL)-1β, and IL-18 and pyroptosis.Missing: growth | Show results with:growth
  78. [78]
    NLRP3 inflammasome-mediated cytokine production and pyroptosis ...
    Apr 12, 2021 · In this review we focus on the role of NLRP3 inflammasome and its components in breast cancer signaling, highlighting that a more detailed ...
  79. [79]
    Overcoming roadblocks to anti-inflammatory NLRP3 inhibitors - Nature
    Sep 18, 2025 · However, clinical trials of MCC950 have been discontinued due to potential hepatoxicity and the compound is ineffective against several NLRP3 ...
  80. [80]
    Discovery of potent and selective inhibitors of human NLRP3 with a ...
    Sep 2, 2025 · Notably, the majority of NLRP3 inhibitors that have advanced to early stage clinical trials are sulfonylureas or MCC950 derivatives (Vande Walle ...Introduction · Results · Discussion · Materials and methods
  81. [81]
    TH5487 specifically targets NLRP3 in FCAS patients resistant to ...
    Jun 9, 2025 · We show that these inhibitors prevent mitochondrial localization of NLRP3 and directly block inflammasome assembly. ... * The Clinical Trials and ...